Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Stiff matrix induces exosome secretion to promote tumour growth

An Author Correction to this article was published on 12 February 2024

This article has been updated

Abstract

Tissue fibrosis and extracellular matrix (ECM) stiffening promote tumour progression. The mechanisms by which ECM regulates its contacting cells have been extensively studied. However, how stiffness influences intercellular communications in the microenvironment for tumour progression remains unknown. Here we report that stiff ECM stimulates the release of exosomes from cancer cells. We delineate a molecular pathway that links stiff ECM to activation of Akt, which in turn promotes GTP loading to Rab8 that drives exosome secretion. We further show that exosomes generated from cells grown on stiff ECM effectively promote tumour growth. Proteomic analysis revealed that the Notch signalling pathway is activated in cells treated with exosomes derived from tumour cells grown on stiff ECM, consistent with our gene expression analysis of liver tissues from patients. Our study reveals a molecular mechanism that regulates exosome secretion and provides insight into how mechanical properties of the ECM control the tumour microenvironment for tumour growth.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Stiff ECM promotes exosome secretion.
Fig. 2: Akt promotes exosome secretion from cells grown on stiff matrix.
Fig. 3: Rab8 regulates stiff ECM-mediated exosome secretion.
Fig. 4: Rabin8 is phosphorylated and activated by Akt in cells grown on stiff ECM.
Fig. 5: Exosomes induced by stiff matrix promote tumour growth.
Fig. 6: Notch signalling is activated in cells grown on stiffness ECM or treated with Exostiff.
Fig. 7: Jagged1 is enriched in Exostiff.

Similar content being viewed by others

Zixuan Zhao, Xinyi Chen, … Hanry Yu

Data availability

Source data are provided with this paper. For patient cohort expression data, the accession code is in the methods section (GSE14520). The gene expression and clinical data for the LCI dataset including 486 tumours and matched non-tumour liver specimens (non-tumour 239 and tumour 247) are available on Gene Expression Omnibus GSE14520 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=gse14520). All other data supporting the findings of this study are available from the corresponding author on reasonable request.

Change history

References

  1. Butcher, D. T., Alliston, T. & Weaver, V. M. A tense situation: forcing tumour progression. Nat. Rev. Cancer 9, 108–122 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Broders-Bondon, F., Nguyen Ho-Bouldoires, T. H., Fernandez-Sanchez, M. E. & Farge, E. Mechanotransduction in tumor progression: the dark side of the force. J. Cell Biol. 217, 1571–1587 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Fattovich, G., Stroffolini, T., Zagni, I. & Donato, F. Hepatocellular carcinoma in cirrhosis: incidence and risk factors. Gastroenterology 127, S35–S50 (2004).

    Article  PubMed  Google Scholar 

  4. Laklai, H. et al. Genotype tunes pancreatic ductal adenocarcinoma tissue tension to induce matricellular fibrosis and tumor progression. Nat. Med. 22, 497–505 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Levental, K. R. et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 139, 891–906 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Mouw, J. K., Ou, G. & Weaver, V. M. Extracellular matrix assembly: a multiscale deconstruction. Nat. Rev. Mol. Cell Biol. 15, 771–785 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Paszek, M. J. et al. Tensional homeostasis and the malignant phenotype. Cancer Cell 8, 241–254 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Thomas, D. & Radhakrishnan, P. Tumor–stromal crosstalk in pancreatic cancer and tissue fibrosis. Mol. Cancer 18, 14 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Haage, A. & Schneider, I. C. Cellular contractility and extracellular matrix stiffness regulate matrix metalloproteinase activity in pancreatic cancer cells. FASEB J. 28, 3589–3599 (2014).

    Article  CAS  PubMed  Google Scholar 

  10. Liu, C. et al. Role of three-dimensional matrix stiffness in regulating the chemoresistance of hepatocellular carcinoma cells. Biotechnol. Appl. Biochem. 62, 556–562 (2015).

    Article  CAS  PubMed  Google Scholar 

  11. Lo, C. M., Wang, H. B., Dembo, M. & Wang, Y. L. Cell movement is guided by the rigidity of the substrate. Biophys. J. 79, 144–152 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Pathak, A. & Kumar, S. Independent regulation of tumor cell migration by matrix stiffness and confinement. Proc. Natl Acad. Sci. USA 109, 10334–10339 (2012).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  13. Plotnikov, S. V. & Waterman, C. M. Guiding cell migration by tugging. Curr. Opin. Cell Biol. 25, 619–626 (2013).

    Article  CAS  PubMed  Google Scholar 

  14. Rice, A. J. et al. Matrix stiffness induces epithelial–mesenchymal transition and promotes chemoresistance in pancreatic cancer cells. Oncogenesis 6, e352 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Swaminathan, V. et al. Mechanical stiffness grades metastatic potential in patient tumor cells and in cancer cell lines. Cancer Res. 71, 5075–5080 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wei, S. C. et al. Matrix stiffness drives epithelial–mesenchymal transition and tumour metastasis through a TWIST1-G3BP2 mechanotransduction pathway. Nat. Cell Biol. 17, 678–688 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Guilluy, C. et al. The Rho GEFs LARG and GEF-H1 regulate the mechanical response to force on integrins. Nat. Cell Biol. 13, 722–727 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Hytonen, V. P. & Wehrle-Haller, B. Mechanosensing in cell-matrix adhesions—converting tension into chemical signals. Exp. Cell. Res. 343, 35–41 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Miranti, C. K. & Brugge, J. S. Sensing the environment: a historical perspective on integrin signal transduction. Nat. Cell Biol. 4, E83–E90 (2002).

    Article  CAS  PubMed  Google Scholar 

  20. Colombo, M., Raposo, G. & Thery, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 30, 255–289 (2014).

    Article  CAS  PubMed  Google Scholar 

  21. Kalluri, R. & LeBleu, V. S. The biology, function, and biomedical applications of exosomes. Science 367, eaau6977 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Pegtel, D. M. & Gould, S. J. Exosomes. Annu. Rev. Biochem. 88, 487–514 (2019).

    Article  CAS  PubMed  Google Scholar 

  23. Mathieu, M., Martin-Jaular, L., Lavieu, G. & Thery, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 21, 9–17 (2019).

    Article  CAS  PubMed  Google Scholar 

  24. Raposo, G. & Stoorvogel, W. Extracellular vesicles: exosomes, microvesicles, and friends. J. Cell Biol. 200, 373–383 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ostrowski, M. et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell Biol. 12, 19–30 (2010).

    Article  CAS  PubMed  Google Scholar 

  26. Becker, A. et al. Extracellular vesicles in cancer: cell-to-cell mediators of metastasis. Cancer Cell 30, 836–848 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kalluri, R. The biology and function of exosomes in cancer. J. Clin. Invest. 126, 1208–1215 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Georges, P. C. et al. Increased stiffness of the rat liver precedes matrix deposition: implications for fibrosis. Am. J. Physiol. Gastrointest. Liver Physiol. 293, G1147–G1154 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Lu, Q. et al. Hepatocellular carcinoma: stiffness value and ratio to discriminate malignant from benign focal liver lesions. Radiology 275, 880–888 (2015).

    Article  PubMed  Google Scholar 

  30. Wang, H. B., Dembo, M., Hanks, S. K. & Wang, Y. L. Focal adhesion kinase is involved in mechanosensing during fibroblast migration. Proc. Natl Acad. Sci. USA 98, 11295–11300 (2001).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  31. Schrader, J. et al. Matrix stiffness modulates proliferation, chemotherapeutic response, and dormancy in hepatocellular carcinoma cells. Hepatology 53, 1192–1205 (2011).

    Article  CAS  PubMed  Google Scholar 

  32. Hattula, K. et al. Characterization of the Rab8-specific membrane traffic route linked to protrusion formation. J. Cell Sci. 119, 4866–4877 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Strom, M., Hume, A. N., Tarafder, A. K., Barkagianni, E. & Seabra, M. C. A family of Rab27-binding proteins. Melanophilin links Rab27a and myosin Va function in melanosome transport. J. Biol. Chem. 277, 25423–25430 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Knodler, A. et al. Coordination of Rab8 and Rab11 in primary ciliogenesis. Proc. Natl Acad. Sci. USA 107, 6346–6351 (2010).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  35. Feng, S. et al. A Rab8 guanine nucleotide exchange factor-effector interaction network regulates primary ciliogenesis. J. Biol. Chem. 287, 15602–15609 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Wang, J. et al. Activation of Rab8 guanine nucleotide exchange factor Rabin8 by ERK1/2 in response to EGF signaling. Proc. Natl Acad. Sci. USA 112, 148–153 (2015).

    Article  CAS  PubMed  ADS  Google Scholar 

  37. Meurette, O. & Mehlen, P. Notch signaling in the tumor microenvironment. Cancer Cell 34, 536–548 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Huang, Q., Li, J., Zheng, J. & Wei, A. The carcinogenic role of the Notch signaling pathway in the development of hepatocellular carcinoma. J. Cancer 10, 1570–1579 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Villanueva, A. et al. Notch signaling is activated in human hepatocellular carcinoma and induces tumor formation in mice. Gastroenterology 143, 1660–1669 (2012).

    Article  CAS  PubMed  Google Scholar 

  40. Ji, J. et al. Hepatic stellate cell and monocyte interaction contributes to poor prognosis in hepatocellular carcinoma. Hepatology 62, 481–495 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Tsuchida, T. & Friedman, S. L. Mechanisms of hepatic stellate cell activation. Nat. Rev. Gastroenterol. Hepatol. 14, 397–411 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Zhou, W. C., Zhang, Q. B. & Qiao, L. Pathogenesis of liver cirrhosis. World J. Gastroenterol. 20, 7312–7324 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Ma, J. H. et al. Mammalian target of rapamycin regulates murine and human cell differentiation through STAT3/p63/Jagged/Notch cascade. J. Clin. Invest. 120, 103–114 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Gonzalez-King, H. et al. Hypoxia inducible factor-1α potentiates Jagged 1-mediated angiogenesis by mesenchymal stem cell-derived exosomes. Stem Cells 35, 1747–1759 (2017).

    Article  CAS  PubMed  Google Scholar 

  45. Rubashkin, M. G. et al. Force engages vinculin and promotes tumor progression by enhancing PI3K activation of phosphatidylinositol (3,4,5)-triphosphate. Cancer Res. 74, 4597–4611 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Mouw, J. K. et al. Tissue mechanics modulate microRNA-dependent PTEN expression to regulate malignant progression. Nat. Med. 20, 360–367 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Walia, V. et al. Akt Regulates a Rab11-Effector Switch Required for Ciliogenesis. Dev. Cell 50, 229–246.e7 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zhang, Y. et al. Progressive and prognosis value of Notch receptors and ligands in hepatocellular carcinoma: a systematic review and meta-analysis. Sci. Rep. 7, 14809 (2017).

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  49. Thery, C., Amigorena, S., Raposo, G. & Clayton, A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr. Protoc. Cell Biol. https://doi.org/10.1002/0471143030.cb0322s30 (2006).

  50. Thery, C. et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 7, 1535750 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Peinado, H. et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 18, 883–891 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Chen, G. et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 560, 382–386 (2018).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  53. Lu, H. Z. et al. PAK signalling drives acquired drug resistance to MAPK inhibitors in BRAF-mutant melanomas. Nature 550, 133–136 (2017).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  54. Patwardhan, A. et al. Routing of the RAB6 secretory pathway towards the lysosome related organelle of melanocytes. Nat. Commun. 8, 15835 (2017).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  55. Goedhart, J. & Luijsterburg, M. S. VolcaNoseR is a web app for creating, exploring, labeling and sharing volcano plots. Sci. Rep. 10, 20560 (2020).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

Download references

Acknowledgements

The work is supported by NIH grants R35 GM141832 to W.G., NCI U01 CA250044 to R.R., V.M.W. and W.G., NCI intramural research programme Z01 BC 010877, Z01 BC 010876, Z01 BC 010313 to H.D. and X.W.W., and Abramson Cancer Center Support Grant (CA016520) to P.A.G.

Author information

Authors and Affiliations

Authors

Contributions

B.W., D.-A.L., L.G. and W.G. conceived the project and designed the experiments. B.W. (Figs. 1a,d, 2a, 3f and 4c,e,f and Extended Data Fig. 1a,c–e), L.G., D.-A.L. (Figs. 1f and 2c,d,k and Extended Data Figs. 1b,f, 2c, 3b,c and 4d), Y.X. and P.K.M. (Figs. 1e,f, 2b, 3e and 6a and Extended Data Fig. 3f) purified and characterized the exosomes. B.W., L.G., D.-A.L., Y.X., L.C. and P.K.M. performed the polyacrylamide gel and cell culture experiments. B.W. (Figs. 3a–d, 4a,b,d, 6b and 7e and Extended Data Figs. 2b and 3a), D.-A.L. (Figs. 1b,c, 2e,h–j, 3g,h and 7a–d and Extended Data Fig. 4a–c), L.G., Z.Y. and Y.X. performed western blot analysis. B.W. and L.G. performed the immunoprecipitation (Fig. 4d–f). B.W. performed the BRET assay, cell proliferation assay, the immunofluorescence (Figs. 2f,g and 3i,j) and electronic microscopy imaging. B.W., L.G., L.C. and T.L. performed the mouse experiments. J.R. performed the GEF assay (Figs. 4g,h). H.D. performed the clinical data analysis. G.B.M. performed the RPPA experiments. J.N. and R.R. performed bioinformatics analyses. B.W., L.G., D.-A.L., Z.Y., H.D., Y.X., E.E.F., X.W.W. and W.G. analysed and interpreted the data. P.A.G. helped with the statistical analysis. B.W. and W.G. wrote the paper. L.G., D.-A.L., Y.X., P.K.M., L.C., S.J., R.G.W., X.W.W., Y.H.C., R.R. and V.M.W. edited the paper. All authors have read and approved the final manuscript.

Corresponding author

Correspondence to Wei Guo.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Jacky G. Goetz, Adam Engler, Marino Zerial and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Characterization of exosomes derived from different cells.

a, Representative TEM image of exosomes purified from the conditioned media of Huh7 cells. Scale bar: 100 nm. b, Purified exosome proteins were quantitated by Bradford assay and the mean was normalized to 1 for exosomal proteins from soft (0.5 kPa) matrix. Exosomes were collected from equal numbers of cells. Values are presented as Mean ± S.D. n = 3. c, The mean diameters of exosomes purified from the conditioned media of Huh7 cells on soft or stiff matrix. Values are presented as mean ± S.D. 3 independent experiments were performed. At least 106 of purified exosomes were measured by NTA for each experiment. d, Exosomes (10 µg) derived from Huh7 cells on soft or stiff matrix were diluted with 1 ml PBS. The particle concentration was determined by NTA. Values are presented as Mean ± S.D. n = 3. e, Exosomes released from the same number of primary hepatocytes grown on matrix with different stiffness were quantified, and the concentration of exosomes released from cells were normalized to the 0.5 kPa group. Values are presented as Mean ± S.D. n = 3. F, Exosomes released from the same number of MCF10A, MCF10AT, and MCF10CA cells grown on matrix with different stiffness were quantified. The amounts of exosomes released from the cells were normalized to the 0.5 kPa for each cell line. Mean ± S.D. n = 3. Source numerical data and unprocessed blots are available in source data. n represents the number of independent experiments.

Source data

Extended Data Fig. 2 Exosome secretion from Huh7 cells treated with Akt and FAK inhibitors.

a, Volcano plot of RPPA data displaying the pattern of protein expression for Huh7 cells cultured on stiff (10 kPa) matrix relative to soft (0.5 kPa) matrix. Significantly up- and down-regulated proteins are indicated by red and blue dots, respectively (cut-off p < 0.05). All the data points were normalized for protein loading and transformed to Log2 values (labeled “NormLog2” on X-axis). b, Western blots showing the up-regulation of p-Akt but not p-ERK in Huh7 cells grown on stiff ECM. Molecular weights (in kDa) are shown to the right. c, Huh7 cells on soft or stiff matrix were treated with DMSO or Akt inhibitor GDC-0068. The conditioned media were collected and proceeded for NTA. Exosome concentration from the cells treated with DMSO on soft matrix was normalized to 1. Values are presented as Mean ± S.D., n = 3. d-f, Huh7 cells were treated with DMSO or various concentrations of Akt inhibitors MK-2206, GDC-0068 or FAK inhibitor PND-1168. Cell viability was examined by CCK-8 assay and normalized to the value of DMSO treated group. Values are presented as Mean ± S.D. n = 3. Source numerical data and unprocessed blots are available in source data. n represents the number of independent experiments.

Source data

Extended Data Fig. 3 Characterization of Rab8 and Rabin8 in cells grown on different matrix.

a, Huh7 cells were transfected with GFP or GFP-Rab8 and grown on soft matrix. Exosomes in the conditioned media were purified by ultracentrifugation, and exosomes from the same number of cells were loaded for western blotting with antibodies against exosome markers HRS and CD63. b, Conditioned media from cells expressing GFP or GFP-Rab8 and treated with DMSO or MK-2206 were collected and proceeded for NTA. Exosome concentration was normalized to those from GFP expressing cells treated with DMSO. Values are presented as Mean ± S.D. n = 3. c, Conditioned media from cells with control or Rab8 shRNA treated with DMSO or MK-2206 were collected and proceeded for NTA. Exosome concentration was normalized to those from cells with control shRNA and DMSO. Values are presented as Mean ± S.D. n = 3. d, Sequence alignment of Rabin8 from different species. The phosphorylation site Serine 149 was shown in red. e, Schematic diagram showing the use of BRET in analyzing Rabin8 conformation. NanoLuc (BRET donor) and HaloTag (BRET acceptor) were fused to the N and C terminus of Rabin8, respectively. When Rabin8 is adopted in a “closed” conformation and autoinhibited, BRET will occur owing to the close proximity between the donor and acceptor. If Rabin8 switches to an “open” conformation induced by Akt phosphorylation on S149, the BRET signal will decrease. f, Huh7 cells were transfected with different Rabin8 variants (WT, S149A, and S149D). Conditioned media were collected and proceeded for NTA. Exosome concentration was normalized to those from cells expressing wild type Rabin8 on soft matrix (n = 3). Values are presented as Mean ± S.D. Source numerical data and unprocessed blots are available in source data. n represents the number of independent experiments.

Source data

Extended Data Fig. 4 Hepa1-6 cells secreted more exosomes when grown on stiff Matrix.

a, Hepa1-6 cells were cultured on soft (0.5 kPa) or stiff (10 kPa) matrix. Exosomes in the conditioned media were purified. Quantification of the exosomal proteins by Bradford assay. The amounts of exosomal proteins were normalized to those from soft matrix. Values are presented as Mean ± S.D. n = 3. b, Exosomes from the same number of cells were analyzed by immunoblotting using antibodies against indicated exosome markers. c, Quantification of the levels of HRS, Syntenin-1, CD63, Alix and Tsg101 is presented. Mean ± S.D. n = 3. d, Exosomes released from the same number of Hepa1-6 cells grown on matrix with different stiffness were quantified, and the concentration of exosomes released from cells grown on 0.5 kPa matrix were normalized as 1. Values are presented as Mean ± S.D. n = 3. n represents the number of independent experiments. e, Growth curves of Hepa1-6 tumors in C57L/J mice injected with PBS or the same amounts of exosomes derived from Hepa1-6 cells treated with DMSO or MK-2206 (n = 5 mice). Values are presented as Mean ± S.D. Source numerical data and unprocessed blots are available in source data.

Source data

Extended Data Fig. 5 Expression of the Notch pathway proteins in Huh7 cells grown on soft or stiff matrix.

Heatmap of RPPA data showing the levels of the Notch pathway proteins in Huh7 cells grown on soft (0.5 kPa) or stiff (10 kPa) matrix. Source numerical data is available in source data.

Source data

Extended Data Fig. 6 Gene expression analysis of liver tissues from patients.

Box and whisker plot (bars represent 10-90 percentile, dots represent outliers) of albumin levels in the serum of 226 HCC patients with high (n = 113 patients) or low (n = 113 patients) Notch gene expression (p value is from Mann-Whitney test (two-tailed). b, Box and whisker plot (bars represent 10-90 percentile, dots represent outliers) of alanine transaminase (ALT) levels in the serum of 226 HCC patients with high (n = 113 patients) or low (n = 113 patients) Notch gene expression (p value is from Mann-Whitney test (two-tailed). c, Analysis of Notch-high and Notch-low patients with HSC or without (nHSC) the HSC gene signature (Fishers Exact test). d, Notch-high (n = 113 patients) and Notch-low (n = 113 patients) patients with or without cirrhosis (Fishers Exact test). The proportion of patients with high Notch genes expression in each group were analyzed (Fishers Exact test). e, Survival risk prediction analysis based on the expression data of four Notch associated genes, HEY1, HEY2, HES1, and SOX9 in non-tumor tissues derived from 226 HCC patients (Kaplan-Meier Cox Log Rank test between two groups and leave one out permutation analyses (1000X)). f, Top enriched signaling pathways associated with Notch activation. Ingenuity Pathway Analysis was performed on 1,872 differentially expressed genes (p < 0.001) between high-Notch) and low-Notch patient non-tumor samples (see Methods for detail). -log(p-value) were calculated using Fisher’s Exact test with Bonferroni correction. g, Box and whisker plot (bars represent 10-90 percentile, dots represent outliers) analysis of Jagged1 expression in 226 HCC patients with Notch-high (n = 113 patients) and Notch-low (n = 113 patients) expression.

Source data

Extended Data Fig. 7 RPPA of the exosomal proteins collected from Huh7 cells grown on soft or stiff matrix.

Volcano plot of RPPA data displaying the pattern of protein expression in exosomes derived from Huh7 cells cultured on stiff relative to soft matrix. Significantly up- and down-regulated proteins are indicated by red and blue dots, respectively (cut-off p < 0.05). All the data points were normalized for protein loading and transformed to Log2 values on X axis. Source numerical data is available in source data.

Source data

Supplementary information

Supplementary Information

Supplementary Table 1. Antibody list.

Reporting Summary

Source data

Source Data Fig. 1

Unprocessed western blots and/or gels.

Source Data Fig. 2

Unprocessed western blots and/or gels.

Source Data Fig. 3

Unprocessed western blots and/or gels.

Source Data Fig. 4

Unprocessed western blots and/or gels.

Source Data Fig. 6

Unprocessed western blots and/or gels.

Source Data Fig. 7

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 2

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 3

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 4

Unprocessed western blots and/or gels.

Source Data Fig. 1

Numeric source data.

Source Data Fig. 2

Numeric source data.

Source Data Fig. 3

Numeric source data.

Source Data Fig. 4

Numeric source data.

Source Data Fig. 5

Numeric source data.

Source Data Fig. 6

Numeric source data.

Source Data Fig. 7

Numeric source data.

Source Data Extended Data Fig. 1

Numeric source data.

Source Data Extended Data Fig. 2

Numeric source data.

Source Data Extended Data Fig. 3

Numeric source data.

Source Data Extended Data Fig. 4

Numeric source data.

Source Data Extended Data Fig. 5

Numeric source data.

Source Data Extended Data Fig. 6

Numeric source data.

Source Data Extended Data Fig. 7

Numeric source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, B., Liu, DA., Guan, L. et al. Stiff matrix induces exosome secretion to promote tumour growth. Nat Cell Biol 25, 415–424 (2023). https://doi.org/10.1038/s41556-023-01092-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-023-01092-1

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer